scholarly journals Depletion of Arg/Abl2 improves endothelial cell adhesion and prevents vascular leak during inflammation

Angiogenesis ◽  
2021 ◽  
Author(s):  
Joana Amado-Azevedo ◽  
Anne-Marieke D. van Stalborch ◽  
Erik T. Valent ◽  
Kalim Nawaz ◽  
Jan van Bezu ◽  
...  

AbstractEndothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.

2008 ◽  
Vol 295 (2) ◽  
pp. L363-L369 ◽  
Author(s):  
Ramaswamy Ramchandran ◽  
Dolly Mehta ◽  
Stephen M. Vogel ◽  
Muhammad K. Mirza ◽  
Panos Kouklis ◽  
...  

Activation of the Rho GTPase Cdc42 has been shown in endothelial cell monolayers to prevent disassembly of interendothelial junctions and the increase in endothelial permeability. Here, we addressed the in vivo role of Cdc42 activity in mediating endothelial barrier protection in lungs by generating mice expressing the dominant active mutant V12Cdc42 protein in vascular endothelial cells targeted via the VE-cadherin promoter. These mice developed normally and exhibited constitutively active GTP-bound Cdc42. The increase in lung vascular permeability and gain in tissue water content in response to intraperitoneal lipopolysaccharide challenge (7 mg/kg) were markedly attenuated in the transgenic mice. To address the basis of the protective effect, we observed that expression of V12Cdc42 mutant in endothelial monolayers reduced the decrease in transendothelial electrical resistance, a measure of opening of interendothelial junctions, thus indicating that Cdc42 activity preserved junctional integrity. RhoA activity in V12Cdc42-expressing endothelial monolayers was reduced compared with untransfected cells, suggesting that activated Cdc42 functions by counteracting the canonical RhoA-mediated mechanism of endothelial hyperpermeability. Therefore, Cdc42 activity of microvessel endothelial cells is a critical determinant of junctional barrier restrictiveness and may represent a means of therapeutically modulating increased lung vascular permeability and edema formation.


Hypertension ◽  
2013 ◽  
Vol 62 (suppl_1) ◽  
Author(s):  
Juan Castor ◽  
Darijana Horvat ◽  
Walter E Cromer ◽  
Thomas J Kuehl ◽  
David C Zawieja ◽  
...  

Objective: Preeclampsia (preE) is a hypertensive disorder unique to pregnancy. Cardiotonic steroids (CTS) such as marinobufagenin (MBG), cinobufotalin (CINO), and ouabain (OUB) are Na + /K + ATPase inhibitors. MBG is elevated in a rat model and patients with preE. MBG causes a vascular leak syndrome in vivo and increases endothelial cell monolayer permeability. Edema is a common syndrome of preE. To assess whether CTS are involved in the leakage of lymphatic endothelial cells (LECs) lining during preE, we evaluated the effect of these CTS on monolayer permeability of LECs in culture. Methods: LECs were isolated from a rat mesenteric collecting lymphatic vessel. The cells were treated with DMSO (vehicle), MBG, CINO, or OUB (1, 10 or 100 nM). Some LECs were pretreated with L-NAME (N-Nitro-L-Arginine Methyl Ester) at a concentration of 1μM before treatment with 100 nM MBG or CINO. Monolayer permeability of CTS-induced LECs was measured by using a fluorescent dye that was quantified on a fluorescence plate reader. The expression of β-catenin and VE-cadherin in the CTS-treated LECs was measured by immunofluorescence. Western blot was performed to measure β-catenin, VE-cadherin, and LYVE-1 protein levels. Statistical comparisons were performed using analysis of variance with Dunnett's post hoc tests. Results: MBG (≥ 1 nM, p<0.05) and CINO (≥ 10 nM, p<0.05) significantly increased the monolayer permeability of LECs compared to DMSO while OUB had no effect. Pretreatment of LECs with 1μM L-NAME attenuated the monolayer permeability of LECs treated with either 100 nM of MBG (p<0.05) or 100 nM of CINO (p<0.05). The β-catenin protein expression in LECs was downregulated by both MBG (p<0.05) and CINO (p<0.05) treatment. However, CTS did not cause any disruption of the LECs tight junctions. CINO (p<0.05) downregulated the VE-cadherin and LYVE-1 protein expression, but MBG did not. Conclusions: We have demonstrated that bufadienolides, MBG and CINO, caused an increase in the monolayer permeability of LECs which was attenuated by L-NAME pretreatment. Moreover, the β-catenin protein expression was downregulated by MBG and CINO treatment with no significant effect on tight junctions. These data suggest that CTS may be involved in the vascular leak syndrome in the LEC lining in preE.


2004 ◽  
Vol 287 (1) ◽  
pp. L153-L167 ◽  
Author(s):  
Alan B. Moy ◽  
Ken Blackwell ◽  
Ning Wang ◽  
Kari Haxhinasto ◽  
Mary K. Kasiske ◽  
...  

The mechanisms of phorbol ester- and thrombin-mediated pulmonary artery endothelial barrier dysfunction were compared. Phorbol ester dibutyrate (PDBU) mediated slow force velocity and less force than thrombin. Taxol did not attenuate PDBU-mediated tension, while it reversed nocodazole-mediated tension. PDBU-mediated tension was not affected by acrylamide; PDBU increased cell stiffness and produced greater declines in transendothelial resistance (TER) than acrylamide. Thus PDBU caused a net increase in tension and did not unload microtubule or intermediate filaments. Microfilament remodeling, determined on the basis of immunocytochemistry and actin solubility, lacked the sensitivity and specificity to predict actin-dependent mechanical properties. Thrombin increased myosin light chain (MLC) kinase site-specific MLC phosphorylation, according to peptide map analysis, whereas PDBU did not increase PKC-specific MLC phosphorylation. The initial PDBU-mediated tension development temporally correlated with PDBU-mediated decline in TER and increased low-molecular-weight caldesmon ( l-CaD) phosphorylation. PDBU-mediated tension development and decreases in TER were associated with a temporal loss of endothelial cell-matrix adhesion, based on a numerical model of TER. Although, on the basis of immunocytochemistry, thrombin-mediated tension was associated with actin insolubility, actin reorganization, and gap formation, these changes did not predict thrombin-mediated gap formation, based on TER and time-lapse differential interference contrast microscopy. These data suggest that PDBU may disrupt endothelial barrier function through loss of cell-matrix adhesion through l-CaD-dependent actin contraction.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 481-481
Author(s):  
Vijay Kumar Reddy Kondreddy ◽  
Usha R. Pendurthi ◽  
Xiao Xu ◽  
John H. Griffin ◽  
L. Vijaya Mohan Rao

Endothelial cell protein C receptor (EPCR) interacts with diverse ligands, in addition to its known ligands protein C and activated protein C (APC). We reported earlier that procoagulant clotting factor VIIa (FVIIa) binds EPCR with the same affinity as APC. FVIIa binding to EPCR leads to the down regulation of the EPCR-mediated anticoagulation pathway. Our recent studies showed that FVIIa, like APC, induces EPCR-dependent cytoprotective signaling through activation of protease activated receptor 1 (PAR1). Recent studies of Griffin, Mosnier and their colleagues revealed that APC noncanonical cleavage of PAR1 at Arg46 site that generates a novel tethered ligand is responsible for APC-induced β-arrestin2-dependent PAR1 biased signaling. It is unknown at present whether FVIIa follows a similar mechanism as APC in inducing PAR1 biased signaling. PAR1 reporter constructs - wild-type and cleavage site-specific mutants - were routinely used to investigate PAR1 cleavage by thrombin, APC, or other proteases, and to determine protease-specific cleavage sites in PAR1. Unfortunately, this approach was not useful in determining any FVIIa cleavage site in PAR1. In contrast to thrombin or APC, FVIIa treatment failed to show a detectable cleavage (over the background) of transfected wild-type PAR1 reporter constructs expressed in cultured endothelial cells. However, in other studies, FVIIa was shown to cleave endogenous PAR1 in endothelial cells as assessed by the loss of cleavage-specific PAR1 mAb binding. The recent generation of transgenic mice strains carrying R41Q or R46Q homozygous point mutations in PAR1 has allowed us in the present study to investigate in vivo mechanisms for PAR1-dependent cytoprotective signaling of FVIIa. We employed two murine injury models, LPS-induced inflammation and VEGF-induced barrier disruption. Murine brain endothelial cells isolated from the PAR1 mutated strains and primary human endothelial cells were used to validate in vivo findings and extend the mechanistic studies. Our studies show that administration of rFVIIa (250 µg/kg body weight) reduced LPS-induced cytokine elaboration and neutrophil infiltration in the lung tissues of wild-type (WT) PAR1 and QQ46-PAR1 mice but not in QQ41-PAR1 mice. Similarly, FVIIa suppression of the VEGF-induced barrier disruption was abolished in the QQ41-PAR1 mice but not in WT and the QQ46-PAR1 mice. Parallel experiments conducted with APC showed, as expected, that it protected WT and QQ41-PAR1 mice but not QQ46-PAR1 mice against LPS-induced inflammation and VEGF-mediated barrier destabilization. In vitro signaling studies performed with brain endothelial cells isolated from WT, QQ41-PAR1 and QQ46-PAR1 mice showed that FVIIa activation of Akt in endothelial cells required Arg41 in PAR1. Additional studies showed that FVIIa-cleaved endogenous PAR1 was readily internalized, whereas APC-cleaved PAR1 remained on the cell surface. Very low concentrations of thrombin (&lt; 1 nM) mimicked FVIIa in inducing PAR1-dependent cytoprotective signaling. However, very low concentration thrombin-induced cytoprotective signaling differed from EPCR-FVIIa-induced cytoprotective signaling in the isoform of ß-arrestin required for the protective effect. EPCR-FVIIa-induced PAR1-mediated cytoprotective signaling was soley mediated via the β-arrestin1-dependent pathway whereas very low dose thrombin-induced cytoprotective effects appear to be mediated by either β-arrestin1 or β-arrestin2. Silencing of ß-arrestin1 or ß-arrestin2 alone did not affect the protective effects of very low doses of thrombin, but the silencing both ß-arrestin1 and ß-arrestin2 together completely prevented a low dose of thrombin-induced protective effect. In summary, our data strongly support the hypothesis that the in vivo mechanism of action for FVIIa's pharmacologic benefits in curbing inflammation and endothelial barrier disruption involves biased signaling of PAR1 due to cleavage at Arg41. Moreover, our studies demonstrate that very low concentrations of thrombin are also capable of inducing PAR1 biased beneficial -cytoprotective signaling by coupling to either ß-arrestin1 or 2. These results emphasize the striking diversity of PAR1's conformational states and interactomes that initiate canonical and biased signaling. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 20-20
Author(s):  
Roger JS Preston ◽  
Jennifer A Johnson ◽  
Fionnuala Ni Ainle ◽  
Shona Harmon ◽  
Owen P. Smith ◽  
...  

Abstract Platelet factor 4 (PF4) is an abundant platelet α-granule chemokine released following platelet activation. PF4 interacts with thrombomodulin and the γ-carboxyglutamic acid (Gla) domain of protein C to significantly enhance activated protein C (APC) generation by the thrombin-thrombomodulin complex on the surface of endothelial cells. However, the protein C Gla domain not only mediates protein C activation in vivo, but also plays a critical role in modulating the diverse functional properties of APC once generated. The functional consequences of the interaction between the APC Gla domain and PF4 in relation to APC anticoagulant, anti-inflammatory and anti-apoptotic functions have not previously been fully defined. In a tissue factor-initiated thrombin generation assay, APC impaired thrombin generation as previously described. However PF4 inhibited APC anticoagulant activity in a concentration-dependent manner (IC50 for PF4 inhibition of APC anticoagulant function, 11μg/ml). In contrast, addition of two other cationic polypeptides protamine and polybrene, both significantly enhanced APC anticoagulant activity in plasma. To elucidate the mechanism through which PF4 inhibits APC anticoagulant activity, we utilized a phospholipid-dependent FVa proteolysis time course assay. In the absence of protein S, PF4 had no effect upon FVa proteolysis by APC, indicating that PF4 does not influence the ability of APC to interact with either anionic phospholipids or FVa. However, in the presence of protein S, PF4 significantly inhibited APC-mediated FVa proteolysis (3–5 fold). Collectively, these findings demonstrate that in addition to enhancing APC generation, PF4 also significantly attenuates APC anticoagulant activity in plasma by impairing critical protein S cofactor enhancement of FVa proteolysis, and suggest that PF4 contributes to the poorly-understood APC resistance phenotype associated with activated platelets. APC bound to the endothelial cell protein C receptor (EPCR) via its Gla domain can activate PAR-1 on endothelial cells, triggering complex intracellular signaling that result in anti-inflammatory and anti-apoptotic cellular responses. To ascertain whether PF4 interaction with the protein C/APC Gla domain might impair APC-EPCR-PAR-1 cytoprotective signaling, APC protection against thrombin-induced endothelial barrier permeability and staurosporine-induced apoptosis in the presence of PF4 was determined. APC significantly attenuated thrombin-induced endothelial cell barrier permeability, as expected. PF4 alone (up to 1μM) had no independent effect upon endothelial barrier permeability, and did not protect against thrombin-mediated increased permeability. In contrast to its inhibition of APC anticoagulant activity, PF4 did not significantly inhibit the endothelial barrier protective properties of APC. To determine whether PF4 might interfere with APC-mediated cytoprotection, staurosporine-induced apoptosis in EAhy926 cells was assessed by RT-PCR quantification of pro-apoptotic (Bax) to anti-apoptotic (Bcl-2) gene expression. Pre-treatment of EAhy926 cells with APC decreased the Bax/Bcl-2 ratio close to that determined for untreated EAhy926 cells. PF4 alone, or in combination with APC, had no effect upon apoptosis-related gene expression as determined by alteration of Bax/Bcl-2 expression ratios in response to staurosporine. In summary, PF4 inhibits APC anticoagulant function via inhibition of essential protein S cofactor enhancement in plasma, whilst retaining EPCR/PAR-1 mediated cytoprotective signalling on endothelial cells. This provides a rationale for how PF4 can exert prothrombotic effects in vivo, but also mediate enhanced APC generation on the surface of endothelial cells to induce both anti-inflammatory and anti-apoptotic events. Based on these observations, we propose that PF4 acts as a critical regulator of APC generation in vivo, but also targets APC towards cytoprotective, rather than anticoagulant functions at sites of vascular injury with concurrent platelet activation.


2020 ◽  
Vol 117 (9) ◽  
pp. 5039-5048 ◽  
Author(s):  
Ying Lin ◽  
Jacob M. Wozniak ◽  
Neil J. Grimsey ◽  
Sravan Girada ◽  
Anand Patwardhan ◽  
...  

Thrombin, a procoagulant protease, cleaves and activates protease-activated receptor-1 (PAR1) to promote inflammatory responses and endothelial dysfunction. In contrast, activated protein C (APC), an anticoagulant protease, activates PAR1 through a distinct cleavage site and promotes anti-inflammatory responses, prosurvival, and endothelial barrier stabilization. The distinct tethered ligands formed through cleavage of PAR1 by thrombin versus APC result in unique active receptor conformations that bias PAR1 signaling. Despite progress in understanding PAR1 biased signaling, the proteins and pathways utilized by thrombin versus APC signaling to induce opposing cellular functions are largely unknown. Here, we report the global phosphoproteome induced by thrombin and APC signaling in endothelial cells with the quantification of 11,266 unique phosphopeptides using multiplexed quantitative mass spectrometry. Our results reveal unique dynamic phosphoproteome profiles of thrombin and APC signaling, an enrichment of associated biological functions, including key modulators of endothelial barrier function, regulators of gene transcription, and specific kinases predicted to mediate PAR1 biased signaling. Using small interfering RNA to deplete a subset of phosphorylated proteins not previously linked to thrombin or APC signaling, a function for afadin and adducin-1 actin binding proteins in thrombin-induced endothelial barrier disruption is unveiled. Afadin depletion resulted in enhanced thrombin-promoted barrier permeability, whereas adducin-1 depletion completely ablated thrombin-induced barrier disruption without compromising p38 signaling. However, loss of adducin-1 blocked APC-induced Akt signaling. These studies define distinct thrombin and APC dynamic signaling profiles and a rich array of proteins and biological pathways that engender PAR1 biased signaling in endothelial cells.


PLoS ONE ◽  
2015 ◽  
Vol 10 (7) ◽  
pp. e0132367 ◽  
Author(s):  
Kohei Ashina ◽  
Yoshiki Tsubosaka ◽  
Tatsuro Nakamura ◽  
Keisuke Omori ◽  
Koji Kobayashi ◽  
...  

2016 ◽  
Vol 311 (2) ◽  
pp. L453-L466 ◽  
Author(s):  
Jurjan Aman ◽  
Ester M. Weijers ◽  
Geerten P. van Nieuw Amerongen ◽  
Asrar B. Malik ◽  
Victor W. M. van Hinsbergh

Despite considerable progress in the understanding of endothelial barrier regulation and the identification of approaches that have the potential to improve endothelial barrier function, no drug- or stem cell-based therapy is presently available to reverse the widespread vascular leak that is observed in acute respiratory distress syndrome (ARDS) and sepsis. The translational gap suggests a need to develop experimental approaches and tools that better mimic the complex environment of the microcirculation in which the vascular leak develops. Recent studies have identified several elements of this microenvironment. Among these are composition and stiffness of the extracellular matrix, fluid shear stress, interaction of endothelial cells (ECs) with pericytes, oxygen tension, and the combination of toxic and mechanic injurious stimuli. Development of novel cell culture techniques that integrate these elements would allow in-depth analysis of EC biology that closely approaches the (patho)physiological conditions in situ. In parallel, techniques to isolate organ-specific ECs, to define EC heterogeneity in its full complexity, and to culture patient-derived ECs from inducible pluripotent stem cells or endothelial progenitor cells are likely to advance the understanding of ARDS and lead to development of therapeutics. This review 1) summarizes the advantages and pitfalls of EC cultures to study vascular leak in ARDS, 2) provides an overview of elements of the microvascular environment that can directly affect endothelial barrier function, and 3) discusses alternative methods to bridge the gap between basic research and clinical application with the intent of improving the translational value of present EC culture approaches.


2019 ◽  
Vol 178 ◽  
pp. 27-36 ◽  
Author(s):  
Maria L. Valle ◽  
Janine Dworshak ◽  
Ashok Sharma ◽  
Ahmed S. Ibrahim ◽  
Mohamed Al-Shabrawey ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document